Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Sci Rep ; 11(1): 7114, 2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33782486

RESUMEN

Prostate-specific membrane antigen (PSMA) is a promising target for the treatment of advanced prostate cancer (PC) and various solid tumors. Although PSMA-targeted radiopharmaceutical therapy (RPT) has enabled significant imaging and prostate-specific antigen (PSA) responses, accumulating clinical data are beginning to reveal certain limitations, including a subgroup of non-responders, relapse, radiation-induced toxicity, and the need for specialized facilities for its administration. To date non-radioactive attempts to leverage PSMA to treat PC with antibodies, nanomedicines or cell-based therapies have met with modest success. We developed a non-radioactive prodrug, SBPD-1, composed of a small-molecule PSMA-targeting moiety, a cancer-selective cleavable linker, and the microtubule inhibitor monomethyl auristatin E (MMAE). SBPD-1 demonstrated high binding affinity to PSMA (Ki = 8.84 nM) and selective cytotoxicity to PSMA-expressing PC cell lines (IC50 = 3.90 nM). SBPD-1 demonstrated a significant survival benefit in two murine models of human PC relative to controls. The highest dose tested did not induce toxicity in immunocompetent mice. The high specific targeting ability of SBPD-1 to PSMA-expressing tumors and its favorable toxicity profile warrant its further development.


Asunto(s)
Aminobenzoatos/farmacología , Oligopéptidos/farmacología , Profármacos/farmacología , Antígeno Prostático Específico/efectos de los fármacos , Aminobenzoatos/administración & dosificación , Aminobenzoatos/toxicidad , Animales , Catepsina B/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/prevención & control , Oligopéptidos/administración & dosificación , Oligopéptidos/toxicidad , Profármacos/administración & dosificación , Profármacos/toxicidad , Neoplasias de la Próstata/patología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Nanotheranostics ; 5(2): 182-196, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33564617

RESUMEN

Enhanced vascular permeability in tumors plays an essential role in nanoparticle delivery. Prostate-specific membrane antigen (PSMA) is overexpressed on the epithelium of aggressive prostate cancers (PCs). Here, we evaluated the feasibility of increasing the delivery of PSMA-targeted magnetic nanoparticles (MNPs) to tumors by enhancing vascular permeability in PSMA(+) PC tumors with PSMA-targeted photodynamic therapy (PDT). Method: PSMA(+) PC3 PIP tumor-bearing mice were given a low-molecular-weight PSMA-targeted photosensitizer and treated with fluorescence image-guided PDT, 4 h after. The mice were then given a PSMA-targeted MNP immediately after PDT and monitored with fluorescence imaging and T2-weighted magnetic resonance imaging (T2-W MRI) 18 h, 42 h, and 66 h after MNP administration. Untreated PSMA(+) PC3 PIP tumor-bearing mice were used as negative controls. Results: An 8-fold increase in the delivery of the PSMA-targeted MNPs was detected using T2-W MRI in the pretreated tumors 42 h after PDT, compared to untreated tumors. Additionally, T2-W MRIs revealed enhanced peripheral intra-tumoral delivery of the PSMA-targeted MNPs. That finding is in keeping with two-photon microscopy, which revealed higher vascular densities at the tumor periphery. Conclusion: These results suggest that PSMA-targeted PDT enhances the delivery of PSMA-targeted MNPs to PSMA(+) tumors by enhancing the vascular permeability of the tumors.


Asunto(s)
Antígenos de Superficie/metabolismo , Glutamato Carboxipeptidasa II/metabolismo , Nanopartículas de Magnetita/administración & dosificación , Fotoquimioterapia , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Línea Celular Tumoral , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Ratones , Fármacos Fotosensibilizantes/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Theranostics ; 10(7): 2888-2896, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194842

RESUMEN

Auger radiopharmaceutical therapy is a promising strategy for micrometastatic disease given high linear energy transfer and short range in tissues, potentially limiting normal tissue toxicities. We previously demonstrated anti-tumor efficacy of a small-molecule Auger electron emitter targeting the prostate-specific membrane antigen (PSMA), 2-[3-[1-carboxy-5-(4-[125I]iodo-benzoylamino)-pentyl]-ureido]-pentanedioic acid), or 125I-DCIBzL, in a mouse xenograft model. Here, we investigated the therapeutic efficacy, long-term toxicity, and biodistribution of 125I-DCIBzL in a micrometastatic model of prostate cancer (PC). Methods: To test the therapeutic efficacy of 125I-DCIBzL in micrometastatic PC, we used a murine model of human metastatic PC in which PSMA+ PC3-ML cells expressing firefly luciferase were injected intravenously in NSG mice to form micrometastatic deposits. One week later, 0, 0.37, 1.85, 3.7, 18.5, 37, or 111 MBq of 125I-DCIBzL was administered (intravenously). Metastatic tumor burden was assessed using bioluminescence imaging (BLI). Long-term toxicity was evaluated via serial weights and urinalysis of non-tumor-bearing mice over a 12-month period, as well as final necropsy. Results: In the micrometastatic PC model, activities of 18.5 MBq 125I-DCIBzL and above significantly delayed development of detectable metastatic disease by BLI and prolonged survival in mice. Gross metastases were detectable in control mice and those treated with 0.37-3.7 MBq 125I-DCIBzL at a median of 2 weeks post-treatment, versus 4 weeks for those treated with 18.5-111 MBq 125I-DCIBzL (P<0.0001 by log-rank test). Similarly, treatment with ≥18.5 MBq 125I-DCIBzL yielded a median survival of 11 weeks, compared with 6 weeks for control mice (P<0.0001). At 12 months, there was no appreciable toxicity via weight, urinalysis, or necropsy evaluation in mice treated with any activity of 125I-DCIBzL, which represents markedly less toxicity than the analogous PSMA-targeted α-particle emitter. Macro-to-microscale dosimetry modeling demonstrated lower absorbed dose in renal cell nuclei versus tumor cell nuclei due to lower levels of drug uptake and cellular internalization in combination with the short range of Auger emissions. Conclusion: PSMA-targeted radiopharmaceutical therapy with the Auger emitter 125I-DCIBzL significantly delayed development of detectable metastatic disease and improved survival in a micrometastatic model of PC, with no long-term toxicities noted at 12 months, suggesting a favorable therapeutic ratio for treatment of micrometastatic PC.


Asunto(s)
Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Radioisótopos de Yodo/administración & dosificación , Glicoproteínas de Membrana/antagonistas & inhibidores , Metástasis de la Neoplasia , Neoplasias de la Próstata , Radiofármacos/uso terapéutico , Animales , Humanos , Masculino , Ratones , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/radioterapia , Células PC-3 , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/radioterapia , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Mol Pharm ; 16(5): 2060-2068, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-30912947

RESUMEN

Magnetic nanoparticle (MNP)-induced hyperthermia is currently being evaluated for localized prostate cancer. We evaluated the feasibility of tumor-selective delivery of prostate-specific membrane antigen (PSMA)-targeted MNPs in a murine model with high-resolution magnetic resonance imaging (MRI) after intravenous administration of MNPs at a concentration necessary for hyperthermia. A PSMA-targeted MNP was synthesized and evaluated using T2-weighted MRI, after intravenous administration of 50 mg/kg of the MNP. Significant contrast enhancement ( P < 0.0002, n = 5) was observed in PSMA(+) tumors compared to PSMA(-) tumors 24 h and 48 h after contrast agent administration. Mice were also imaged with near-infrared fluorescence imaging, to validate the MRI results. Two-photon microscopy revealed higher vascular density at the tumor periphery, which resulted in higher  peripheral accumulation of PSMA-targeted MNPs. These results suggest that the delivery of PSMA-targeted MNPs to PSMA(+) tumors is both actively targeted and passively mediated.


Asunto(s)
Antígenos de Superficie/química , Antígenos de Superficie/metabolismo , Medios de Contraste/química , Glutamato Carboxipeptidasa II/química , Glutamato Carboxipeptidasa II/metabolismo , Imagen por Resonancia Magnética , Nanopartículas de Magnetita/química , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/terapia , Animales , Antígenos de Superficie/genética , Estudios de Cohortes , Sistemas de Liberación de Medicamentos , Estudios de Factibilidad , Glutamato Carboxipeptidasa II/genética , Humanos , Hipertermia Inducida , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Imagen Óptica , Células PC-3 , Transfección , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Biomacromolecules ; 18(1): 201-209, 2017 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-28001364

RESUMEN

Targeted delivery of drug-encapsulated nanoparticles is a promising new approach to safe and effective therapeutics for cancer. Here we investigate the pharmacokinetics and biodistribution of a prostate-specific membrane antigen (PSMA)-targeted nanoparticle based on a poly(lactic acid)-polyethylene glycol copolymer by utilizing single photon emission computed tomography (SPECT) and fluorescence imaging of a low-molecular-weight, PSMA-targeting moiety attached to the surface and oriented toward the outside environment. Tissue biodistribution of the radioactive, PSMA-targeted nanoparticles in mice containing PSMA(+) PC3 PIP and PSMA(-) PC3 flu (control) tumors demonstrated similar accumulation compared to the untargeted particles within all tissues except for the tumor and liver by 96 h postinjection. For PSMA(+) PC3 PIP tumor, the targeted nanoparticle demonstrated retention of 6.58% injected dose (ID)/g at 48 h and remained nearly at that level out to 96 h, whereas the untargeted nanoparticle showed a 48 h retention of 8.17% ID/g followed by a significant clearance to 2.37% ID/g at 96 h (P < 0.02). On the other hand, for control tumor, both targeted and untargeted particles displayed similar 48 h retentions and rates of clearance over 96 h. Ex vivo microscopic analysis with near-infrared versions of the nanoparticles indicated retention within PSMA(+) tumor epithelial cells as well as tumor-associated macrophages for targeted particles and primarily macrophage-associated uptake for the untargeted particles. Retention in control tumor was primarily associated with tumor vasculature and macrophages. The data demonstrate the utility of radioimaging to assess nanoparticle biodistribution and suggest that active targeting has a modest positive effect on tumor localization of PSMA-targeted PLA-PEG nanoparticles that have been derivatized for imaging.


Asunto(s)
Antígenos de Superficie/metabolismo , Bencenosulfonatos/farmacocinética , Glutamato Carboxipeptidasa II/metabolismo , Radioisótopos de Indio/farmacocinética , Indoles/farmacocinética , Nanopartículas/administración & dosificación , Polietilenglicoles/química , Neoplasias de la Próstata/diagnóstico por imagen , Radiofármacos/farmacocinética , Animales , Colorantes Fluorescentes/farmacocinética , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Nanopartículas/química , Polímeros/administración & dosificación , Polímeros/química , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Espectroscopía Infrarroja Corta , Distribución Tisular , Células Tumorales Cultivadas
6.
Biomaterials ; 80: 57-67, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26706476

RESUMEN

Metastatic prostate cancer causes significant morbidity and mortality and there is a critical unmet need for effective treatments. We have developed a theranostic nanoplex platform for combined imaging and therapy of prostate cancer. Our prostate-specific membrane antigen (PSMA) targeted nanoplex is designed to deliver plasmid DNA encoding tumor necrosis factor related apoptosis-inducing ligand (TRAIL), together with bacterial cytosine deaminase (bCD) as a prodrug enzyme. Nanoplex specificity was tested using two variants of human PC3 prostate cancer cells in culture and in tumor xenografts, one with high PSMA expression and the other with negligible expression levels. The expression of EGFP-TRAIL was demonstrated by fluorescence optical imaging and real-time PCR. Noninvasive (19)F MR spectroscopy detected the conversion of the nontoxic prodrug 5-fluorocytosine (5-FC) to cytotoxic 5-fluorouracil (5-FU) by bCD. The combination strategy of TRAIL gene and 5-FC/bCD therapy showed significant inhibition of the growth of prostate cancer cells and tumors. These data demonstrate that the PSMA-specific theranostic nanoplex can deliver gene therapy and prodrug enzyme therapy concurrently for precision medicine in metastatic prostate cancer.


Asunto(s)
Antimetabolitos/administración & dosificación , ADN/administración & dosificación , Sistemas de Liberación de Medicamentos , Flucitosina/administración & dosificación , Profármacos/administración & dosificación , Neoplasias de la Próstata/terapia , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Animales , Antígenos de Superficie/metabolismo , Antimetabolitos/metabolismo , Antimetabolitos/uso terapéutico , Bacterias/enzimología , Línea Celular Tumoral , Citosina Desaminasa/administración & dosificación , Citosina Desaminasa/metabolismo , Citosina Desaminasa/uso terapéutico , ADN/genética , ADN/uso terapéutico , Terapia Enzimática , Flucitosina/metabolismo , Flucitosina/uso terapéutico , Terapia Genética , Glutamato Carboxipeptidasa II/metabolismo , Humanos , Masculino , Ratones , Modelos Moleculares , Plásmidos/administración & dosificación , Plásmidos/genética , Plásmidos/uso terapéutico , Profármacos/metabolismo , Profármacos/uso terapéutico , Próstata/efectos de los fármacos , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Nanomedicina Teranóstica
7.
Nanoscale ; 7(10): 4432-42, 2015 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-25675333

RESUMEN

Early detection enables improved prognosis for prostate cancer (PCa). A promising target for imaging and therapy of PCa is the prostate-specific membrane antigen (PSMA), which exhibits both expression within the epithelium of PCa cells, and becomes internalized upon ligand binding. Here we report the synthesis of a PSMA-targeted bionized nanoferrite (BNF) nanoparticle and its biological evaluation in an experimental model of PCa. The BNF nanoparticle formulation exhibits properties conducive to targeted imaging such as stealth, prolonged circulation time and enhanced clearance from non-target sites. Optical imaging of the targeted BNF in vivo indicates preferential accumulation in PSMA+ tumors 4 h post-injection, suggesting target specificity. On the other hand, non-targeted nanoparticles exhibit lower uptake with similar accumulation in both PSMA+ and PSMA- tumors indicating tumor access without preferential accumulation. Imaging with single photon emission computed tomography (SPECT) and biodistribution studies of a modified construct indicate highest tumor accumulation at 48 h post-injection [4.3 ± 0.4 percentage injected dose per gram of tissue (%ID g(-1))], with tumor/blood and tumor/muscle ratios of 7.5 ± 2.4 and 11.6 ± 1.2 %ID g(-1), respectively. Ex vivo fluorescence microscopy, Prussian blue staining, immunohistochemistry and biodistribution studies confirm enhanced nanoparticle uptake in PSMA+ tumors compared to those not expressing PSMA. The BNF nano-formulation described is promising for PSMA-targeted imaging applications in vivo.


Asunto(s)
Antígenos de Superficie/metabolismo , Antineoplásicos , Sistemas de Liberación de Medicamentos/métodos , Compuestos Férricos , Glutamato Carboxipeptidasa II/metabolismo , Nanopartículas/química , Neoplasias de la Próstata , Tomografía Computarizada de Emisión de Fotón Único , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Compuestos Férricos/química , Compuestos Férricos/farmacología , Humanos , Masculino , Ratones , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Radiografía , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Bioconjug Chem ; 25(2): 393-405, 2014 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-24410012

RESUMEN

Differential expression of surface proteins on normal vs malignant cells provides the rationale for the development of receptor-, antigen-, and transporter-based, cancer-selective imaging and therapeutic agents. However, tumors are heterogeneous, and do not always express what can be considered reliable, tumor-selective markers. That suggests development of more flexible targeting platforms that incorporate multiple moieties enabling concurrent targeting to a variety of putative markers. We report the synthesis, biochemical, in vitro, and preliminary in vivo evaluation of a new heterobivalent (HtBv) imaging agent targeting both the prostate-specific membrane antigen (PSMA) and integrin-αvß3 surface markers, each of which can be overexpressed in certain tumor epithelium and/or neovasculature. The HtBv agent was functionalized with either 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) or the commercially available IRDye800CW. DOTA-conjugated HtBv probe 9 bound to PSMA or αvß3 with affinities similar to those of monovalent (Mnv) compounds designed to bind to their targets independently. In situ energy minimization experiments support a model describing the conformations adapted by 9 that enable it to bind both targets. IRDye800-conjugated HtBv probe 10 demonstrated target-specific binding to either PSMA or integrin-αvß3 overexpressing xenografts. HtBv agents 9 and 10 may enable dual-targeted imaging of malignant cells and tissues in an effort to address heterogeneity that confounds many cancer-targeted imaging agents.


Asunto(s)
Antígenos de Superficie/química , Glutamato Carboxipeptidasa II/química , Integrina alfaVbeta3/química
9.
Bioconjug Chem ; 23(12): 2377-85, 2012 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-23157641

RESUMEN

Targeted near-infrared (NIR) optical imaging can be used in vivo to detect specific tissues, including malignant cells. A series of NIR fluorescent ligands targeting the prostate-specific membrane antigen (PSMA) was synthesized and each compound was tested for its ability to image PSMA+ tissues in experimental models of prostate cancer. The agents were prepared by conjugating commercially available active esters of NIR dyes, including IRDye800CW, IRDye800RS, Cy5.5, Cy7, or a derivative of indocyanine green (ICG) to the terminal amine group of (S)-2-(3-((S)-5-amino-1-carboxypentyl)ureido)pentanedioic acid 1, (14S,18S)-1-amino-8,16-dioxo-3,6-dioxa-9,15,17-triazaicosane-14,18,20-tricarboxylic acid 2 and (3S,7S)-26-amino-5,13,20-trioxo-4,6,12,21-tetraazahexacosane-1,3,7,22-tetracarboxylic acid 3. The K(i) values for the dye-inhibitor conjugates ranged from 1 to 700 pM. All compounds proved capable of imaging PSMA+ tumors selectively to varying degrees depending on the choice of fluorophore and linker. The highest tumor uptake was observed with IRDye800CW employing a poly(ethylene glycol) or lysine-suberate linker, as in 800CW-2 and 800CW-3, while the highest tumor to nontarget tissue ratios were obtained for Cy7 with these same linkers, as in Cy7-2 and Cy7-3. Compounds 2 and 3 provide useful scaffolds for targeting of PSMA+ tissues in vivo and should be useful for preparing NIR dye conjugates designed specifically for clinical intraoperative optical imaging devices.


Asunto(s)
Colorantes Fluorescentes/síntesis química , Glutamato Carboxipeptidasa II/análisis , Glicoproteínas de Membrana/análisis , Imagen Molecular/métodos , Próstata/patología , Neoplasias de la Próstata/patología , Espectroscopía Infrarroja Corta/métodos , Animales , Ácidos Carboxílicos/química , Modelos Animales de Enfermedad , Colorantes Fluorescentes/farmacocinética , Colorantes Fluorescentes/farmacología , Glutamato Carboxipeptidasa II/metabolismo , Humanos , Cinética , Lisina/análogos & derivados , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Desnudos , Imagen Molecular/instrumentación , Peso Molecular , Polietilenglicoles/química , Próstata/efectos de los fármacos , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Espectroscopía Infrarroja Corta/instrumentación
10.
ACS Nano ; 6(9): 7752-7762, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-22866897

RESUMEN

Theranostic imaging, where diagnosis is combined with therapy, is particularly suitable for a disease that is as complex as cancer, especially now that genomic and proteomic profiling can provide an extensive "fingerprint" of each tumor. With such information, theranostic agents can be designed to personalize treatment and minimize damage to normal tissue. Here we have developed a nanoplex platform for theranostic imaging of prostate cancer (PCa). In these proof-of-principle studies, a therapeutic nanoplex containing multimodal imaging reporters was targeted to prostate-specific membrane antigen (PSMA), which is expressed on the cell surface of castrate-resistant PCa. The nanoplex was designed to deliver small interfering RNA (siRNA) along with a prodrug enzyme to PSMA-expressing tumors. Each component of the nanoplex was carefully selected to evaluate its diagnostic aspect of PSMA imaging and its therapeutic aspects of siRNA-mediated down-regulation of a target gene and the conversion of a prodrug to cytotoxic drug, using noninvasive multimodality imaging. Studies performed using two variants of human PC3-PCa cells and tumors, one with high PSMA expression level and another with negligible expression levels, demonstrated PSMA-specific uptake. In addition, down-regulation of the selected siRNA target, choline kinase (Chk), and the conversion of the nontoxic prodrug 5-fluorocytosine (5-FC) to cytotoxic 5-fluorouracil (5-FU) were also demonstrated with noninvasive imaging. The nanoplex was well-tolerated and did not induce liver or kidney toxicity or a significant immune response. The nanoplex platform described can be easily modified and applied to different cancers, receptors, and pathways to achieve theranostic imaging, as a single agent or in combination with other treatment modalities.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/uso terapéutico , Antígeno Prostático Específico/química , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/terapia , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Nanopartículas/química , Especificidad de Órganos , Neoplasias de la Próstata/metabolismo , Distribución Tisular , Resultado del Tratamiento
11.
Mol Ther ; 19(9): 1626-35, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21750533

RESUMEN

Successful translation of nonviral gene delivery to therapeutic applications requires detailed understanding of in vivo trafficking of the vehicles. This report compares the pharmacokinetic and biodistribution profiles of polyethylene glycol-b-polyphosphoramidate (PEG-b-PPA)/DNA micellar nanoparticles after administration through intravenous infusion, intrabiliary infusion, and hydrodynamic injection using single photon emission computed tomography/computed tomography (SPECT/CT) imaging. Nanoparticles were labeled with (111)In using an optimized protocol to retain their favorable physicochemical properties. Quantitative imaging analysis revealed different in vivo trafficking kinetics for PEG-b-PPA/DNA nanoparticles after different routes of administration. The intrabiliary infusion resulted in the highest liver uptake of micelles compared with the other two routes. Analysis of intrabiliary infusion by the two-compartment pharmacokinetic modeling revealed efficient retention of micelles in the liver and minimal micelle leakage from the liver to the blood stream. This study demonstrates the utility of SPECT/CT as an effective noninvasive imaging modality for the characterization of nanoparticle trafficking in vivo and confirms that intrabiliary infusion is an effective route for liver-targeted delivery of DNA-containing nanoparticles.


Asunto(s)
ADN/química , Micelas , Nanopartículas/química , Plásmidos/administración & dosificación , Polietilenglicoles/química , Tomografía Computarizada de Emisión de Fotón Único/métodos , Animales , Fenómenos Químicos , Femenino , Técnicas de Transferencia de Gen , Infusiones Intravenosas , Hígado , Nanopartículas/administración & dosificación , Plásmidos/química , Polietilenglicoles/farmacocinética , Ratas , Ratas Wistar
12.
Bioconjug Chem ; 20(8): 1625-33, 2009 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-19572702

RESUMEN

Technetium tricarbonyl chemistry has been a subject of interest in radiopharmaceutical development over the past decade. Despite the extensive work done on developing chelates for Tc(I), a rigorous investigation of the impact of changing donor groups and labeling conditions on radiochemical yields and/or distribution has been lacking. This information is crucially important if these platforms are going to be used to develop molecular imaging probes. Previous studies on the coordination chemistry of the {M(CO)(3)}(+) core have established alkylamine, aromatic nitrogen heterocycles, and carboxylate donors as effective chelating ligands. These observations led to the design of tridentate ligands derived from the amino acid lysine. Such amino acid analogues provide a tridentate donor set for chelation to the metal and an amino acid functionality for conjugation to biomolecules. We recently developed a family of single amino acid chelates (SAAC) that serve this function and can be readily incorporated into peptides via solid-phase synthesis techniques. As part of these continuing studies, we report here on the radiolabeling with technetium-99m ((99m)Tc) and stability of a series of SAAC analogues of lysine. The complexes studied include cationic, neutral, and anionic complexes. The results of tissue distribution studies with these novel complexes in normal rats demonstrate a range of distribution in kidney, liver, and intestines.


Asunto(s)
Aminoácidos/farmacocinética , Quelantes/farmacocinética , Lisina/farmacología , Radiofármacos/farmacocinética , Tecnecio/farmacocinética , Aminoácidos/química , Animales , Quelantes/química , Mucosa Intestinal/metabolismo , Marcaje Isotópico , Riñón/metabolismo , Hígado/metabolismo , Lisina/análogos & derivados , Lisina/química , Masculino , Estructura Molecular , Radiofármacos/química , Ratas , Ratas Sprague-Dawley , Estereoisomerismo , Tecnecio/química , Distribución Tisular
13.
Bioorg Med Chem ; 17(14): 4888-93, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19541487

RESUMEN

Prostate-specific antigen (PSA) is a serine protease biomarker that may play a role in prostate cancer development and progression. The inhibition of PSA's enzymatic activity with small molecule inhibitors is an attractive and, as of yet, unexploited target. Previously, we reported a series of peptidyl aldehyde and boronic acid based inhibitors of PSA. In this study, the structural requirements in the P2 and P3 positions of peptide-based PSA inhibitors are explored through the substitution of a series of natural and unnatural amino acids in these positions. This analysis demonstrated a preference for hydrophobic residues in the P2 position and amino acids with the potential to hydrogen bond in the P3 position. Using this information, a peptide boronic acid inhibitor with the sequence Cbz-Ser-Ser-Gln-Nle-(boro)-Leu was identified with a K(i) for PSA of 25nM. The attachment of a bulky metal chelating group to the amino terminal of this peptide did not adversely affect PSA inhibition. This result suggests that a platform of PSA inhibitor chelates could be developed as SPECT or PET-based imaging agents for prostate cancer.


Asunto(s)
Péptidos/química , Péptidos/farmacología , Antígeno Prostático Específico/antagonistas & inhibidores , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/enzimología , Secuencia de Aminoácidos , Ácidos Borónicos/síntesis química , Ácidos Borónicos/química , Ácidos Borónicos/farmacología , Diagnóstico por Imagen , Humanos , Masculino , Biblioteca de Péptidos , Péptidos/síntesis química , Antígeno Prostático Específico/aislamiento & purificación , Semen/química , Especificidad por Sustrato
14.
J Med Chem ; 51(24): 7737-43, 2008 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-19053759

RESUMEN

Urea-based, low molecular weight ligands of glutamate carboxypeptidase II (GCPII) have demonstrated efficacy in various models of neurological disorders and can serve as imaging agents for prostate cancer. To enhance further development of such compounds, we determined X-ray structures of four complexes between human GCPII and urea-based inhibitors at high resolution. All ligands demonstrate an invariant glutarate moiety within the S1' pocket of the enzyme. The ureido linkage between P1 and P1' inhibitor sites interacts with the active-site Zn(1)(2+) ion and the side chains of Tyr552 and His553. Interactions within the S1 pocket are defined primarily by a network of hydrogen bonds between the P1 carboxylate group of the inhibitors and the side chains of Arg534, Arg536, and Asn519. Importantly, we have identified a hydrophobic pocket accessory to the S1 site that can be exploited for structure-based design of novel GCPII inhibitors with increased lipophilicity.


Asunto(s)
Glutamato Carboxipeptidasa II/química , Urea/farmacología , Zinc/química , Sitios de Unión , Química Farmacéutica/métodos , Diseño de Fármacos , Ácido Glutámico/química , Humanos , Iones , Ligandos , Modelos Moleculares , Conformación Molecular , Conformación Proteica , Estructura Terciaria de Proteína , Tirosina/química , Urea/química
15.
Cancer Biol Ther ; 7(6): 974-82, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18698158

RESUMEN

Polymeric nanoparticles represent a form of targeted therapy due to their ability to passively accumulate within the tumor interstitium via the enhanced permeability and retention (EPR) effect. We used a combined approach to decorate the surface of a nanoparticle with a urea-based small-molecule peptidomimetic inhibitor of prostate specific membrane antigen (PSMA). PSMA is expressed by normal and malignant prostate epithelial cells and by the neovasculature of almost all solid tumors. This strategy takes advantage of both the avidity of the functionalized nanoparticle for binding to PSMA and the ability of the nanoparticle to be retained for longer periods of time in the tumor due to enhanced leakage via EPR into the tumor interstitium. As an initial step to introducing the targeting moiety, the amino terminus of the small-molecule PSMA inhibitor was conjugated to PEG (M(n) 3400) bearing an activated carboxyl group to obtain a PEGylated inhibitor. Studies undertaken using a radiolabeled PSMA-substrate based assay established that the PEGylated inhibitor had an IC(50) value similar to the uncomplexed inhibitor. Subsequently, nanoparticles loaded with docetaxel were formulated using a mixture of poly(lactide-beta-ethylene glycol-beta-lactide) and PSMA-inhibitor bound alpha-amino-omega-hydroxy terminated poly (ethylene glycol-beta-epsilon-caprolactone). In vitro studies using these nanoparticles demonstrated selective cytotoxicity against PSMA-producing cells. Binding of fluorescently labeled PSMA-targeted particles to PSMA-producing cells has also been directly observed using fluorescence microscopy and observed in secondary fashion using a PSMA substrate based enzyme inhibition assay. Ongoing in vivo studies address the localization, activity and toxicity of these targeted nanoparticles against PSMA-producing human prostate tumor xenografts.


Asunto(s)
Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Nanopartículas/química , Nanotecnología/métodos , Urea/química , Antígenos de Neoplasias/química , Antígenos de Superficie , Línea Celular Tumoral , Endocitosis , Matriz Extracelular/metabolismo , Humanos , Concentración 50 Inhibidora , Masculino , Microscopía Fluorescente/métodos , Polietilenglicoles/química , Polímeros/química , Unión Proteica
16.
J Med Chem ; 51(15): 4504-17, 2008 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-18637669

RESUMEN

The prostate-specific membrane antigen (PSMA) is increasingly recognized as a viable target for imaging and therapy of cancer. We prepared seven (99m)Tc/Re-labeled compounds by attaching known Tc/Re chelating agents to an amino-functionalized PSMA inhibitor (lys-NHCONH-glu) with or without a variable length linker moiety. K i values ranged from 0.17 to 199 nM. Ex vivo biodistribution and in vivo imaging demonstrated the degree of specific binding to engineered PSMA+ PC3 PIP tumors. PC3-PIP cells are derived from PC3 that have been transduced with the gene for PSMA. Despite demonstrating nearly the lowest PSMA inhibitory potency of this series, [(99m)Tc(CO)3( L1)] (+) ( L1 = (2-pyridylmethyl)2N(CH2) 4CH(CO2H)NHCO-(CH2) 6CO-NH-lys-NHCONH-glu) showed the highest, most selective PIP tumor uptake, at 7.9 +/- 4.0% injected dose per gram of tissue at 30 min postinjection. Radioactivity cleared from nontarget tissues to produce a PIP to flu (PSMA-PC3) ratio of 44:1 at 120 min postinjection. PSMA can accommodate the steric requirements of (99m)Tc/Re complexes within PSMA inhibitors, the best results achieved with a linker moiety between the epsilon amine of the urea lysine and the chelator.


Asunto(s)
Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Animales , Sitios de Unión , Línea Celular Tumoral , Quelantes/síntesis química , Quelantes/química , Electrones , Glutamato Carboxipeptidasa II/metabolismo , Masculino , Ratones , Estructura Molecular , Trasplante de Neoplasias , Unión Proteica , Renio/química , Relación Estructura-Actividad , Tecnecio/química , Urea/química
17.
Bioconjug Chem ; 16(4): 885-902, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16029030

RESUMEN

Hydrazinonicotinamide (HYNIC) forms stable coordination complexes with Tc-99m when reacted with Tc(V)oxo species such as Tc-mannitol or other Tc-polyhydric complexes. However, radio-HPLC of [Tc-For-MLFK-HYNIC] labeled via Tc-polyhydric ligands demonstrated multiple radiochemical species each with unique biodistribution patterns. This is likely due to the fact that Tc can bind to the hydrazino moiety, as well as polyhydric ligands, in a variety of coordination geometries. Tridentate ligands, such as bis(mercaptoethyl)methylamine (NS2), may constrain the possible coordination geometries and improve overall stability. To investigate this, we synthesized NS2, converted the [Tc-mannitol-For-MLFK-HYNIC] to the corresponding NS2-containing complex [Tc-NS2-For-MLFK-HYNIC], and compared its infection imaging and biodistribution properties with [Tc-mannitol-For-MLFK-HYNIC]. Conversion to the NS2 complex was confirmed by HPLC which showed a single unique hydrophobic species with retention time greater than the [Tc-mannitol-For-MLFK-HYNIC] complex. Imaging experiments with both preparations were performed in rabbits with E. coli infections in the left thigh. Tissue radioactivity measurements demonstrated that compared to Tc-mannitol-peptide, accumulation of Tc-NS2-peptide was lower in blood, heart, and normal muscle and higher in spleen, infected muscle, and pus (p < 0.01). These results indicate that the Tc-NS2-peptide complex is chemically more homogeneous and exhibits improved infection localization and biodistribution properties. In an effort to model the interactions of the metal-HYNIC core with NS2 and related ligand types, the reactions of [ReCl3(NNC5H4NH)(NHNC5H4N)] and [99TcCl3(NNC5H4NH)(NHNC5H4N)], effective structural analogues for the [M(NNC5H4NH(x))2] core, with NS2, C5H3N-2,6-(CH2SH)2, O(CH2CH2SH)2, and S(CH2CH2SH)2 were investigated and the compounds [M[CH3N(CH2CH2S)2](NNC5H4N)(NHNC5H4N] (M = 99Tc (5a), Re (5b)), [Re[C5H3N-2,6-(CH2S)2](NNC5H4N)(NHNC5H4N)].CH2Cl2.0.5MeOH (7), [Re[SCH2CH2)2O] (NNC5H4N)(NHNC5H4N)] (8), and [Re[(SCH2CH2)2S](NNC5H4NH)(NHNC5H4N)]Cl (9) were isolated. Similarly, the reaction of [ReCl3(NNC5H4NH)(NHNC5H4N)] with the bidentate ligands pyridine-2-methanethiol and 3-(trimethlysilyl)pyridine-2-thiol led to the isolation of [ReCl(C5H4N-2-CH2S) (NNC5H4N)(NHNC5H4N)] (10) and [Re(2-SC5H3N-3-SiMe3)2 (NNC5H4N)(NHNC5H4N)] (11), respectively, while reaction with N-methylimidazole-2-thiol yielded the binuclear complex [Re(OH)Cl(SC3H2N2CH3)2(NNC5H4N)2 (NHNC5H4N)2] (12). The analogous metal-(HYNIC-OH) precursor, [ReCl3[NNC5H3NH(CO2R)] [NHNC5H3N(CO2R)]] (R = H, 13a; R = CH3, 13b) has been prepared and coupled to lysine to provide [RCl3[NNC5H3NH(CONHCH2CH2CH2CH2CH(NH2)CO2H)] [NHNC5H3NH(CONHCH2CH2CH2CH2CH(NH2)CO2H)]].2HCl (14.2HCl), while the reaction of the methyl ester 13b with 2-mercaptopyridine yields [Re(2-SC5H4N)2[NNC5H3N(CO2Me)][NHNC5H3N(CO2Me)]] (15). While the chemical studies confirm the robustness of the M-HYNIC core (M = Tc, Re) and its persistence in ligand substitution reactions at adjacent coordination sites of the metal, the isolation of oligomeric structures and the insolubility of the peptide conjugates of 13, 14, and 15 underscore the difficulty of characterizing these materials on the macroscopic scale, an observation relevant to the persistent concerns with reagent purity and identity on the tracer level.


Asunto(s)
Hidrazinas/química , Mercaptoetilaminas/química , Niacinamida/análogos & derivados , Compuestos de Organotecnecio/química , Péptidos/química , Animales , Cromatografía Líquida de Alta Presión , Cristalografía por Rayos X , Hidrazinas/farmacocinética , Ligandos , Espectroscopía de Resonancia Magnética , Masculino , Modelos Moleculares , Estructura Molecular , Niacinamida/química , Niacinamida/farmacocinética , Compuestos de Organotecnecio/farmacocinética , Péptidos/farmacocinética , Conejos , Espectrofotometría Infrarroja , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA